1
|
Bery AI, Belousova N, Hachem RR, Roux A, Kreisel D. Chronic Lung Allograft Dysfunction: Clinical Manifestations and Immunologic Mechanisms. Transplantation 2025; 109:454-466. [PMID: 39104003 PMCID: PMC11799353 DOI: 10.1097/tp.0000000000005162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/07/2024]
Abstract
The term "chronic lung allograft dysfunction" has emerged to describe the clinical syndrome of progressive, largely irreversible dysfunction of pulmonary allografts. This umbrella term comprises 2 major clinical phenotypes: bronchiolitis obliterans syndrome and restrictive allograft syndrome. Here, we discuss the clinical manifestations, diagnostic challenges, and potential therapeutic avenues to address this major barrier to improved long-term outcomes. In addition, we review the immunologic mechanisms thought to propagate each phenotype of chronic lung allograft dysfunction, discuss the various models used to study this process, describe potential therapeutic targets, and identify key unknowns that must be evaluated by future research strategies.
Collapse
Affiliation(s)
- Amit I Bery
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Natalia Belousova
- Pneumology, Adult Cystic Fibrosis Center and Lung Transplantation Department, Foch Hospital, Suresnes, France
| | - Ramsey R Hachem
- Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Antoine Roux
- Pneumology, Adult Cystic Fibrosis Center and Lung Transplantation Department, Foch Hospital, Suresnes, France
- Paris Transplant Group, INSERM U 970s, Paris, France
| | - Daniel Kreisel
- Departments of Surgery, Pathology & Immunology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
2
|
Moghbeli K, Lipp MA, Bueno M, Craig A, Rojas M, Abbas M, Lakkis ZI, Chuan B, Sembrat J, Noda K, Kass DJ, Chen K, Fan L, Oury T, Zhou Z, Wang X, McDyer JF, Eickelberg O, Snyder ME. NKG2D blockade impairs tissue-resident memory T cell accumulation and reduces chronic lung allograft dysfunction. JCI Insight 2025; 10:e184048. [PMID: 39989456 PMCID: PMC11949055 DOI: 10.1172/jci.insight.184048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 01/09/2025] [Indexed: 02/25/2025] Open
Abstract
Chronic lung allograft dysfunction (CLAD) substantially limits long-term survival following lung transplantation. To identify potential targets for CLAD prevention, T cells from explanted CLAD lungs and lung-draining lymph nodes, as well as diseased and nondiseased controls were isolated and single-cell RNA sequencing and TCR sequencing were performed. TCR sequencing revealed a clonally expanded population of CD8+ tissue-resident memory T cells (TRMs) with high cytotoxic potential, including upregulation of KLRK1, encoding the co-receptor NKG2D. These cytotoxic CD8+ TRMs accumulated around the CLAD airways and had a 100-fold increase in clonal overlap with lung-draining lymph nodes when compared with non-CLAD lungs. Using a murine model of orthotopic lung transplantation, we confirmed that cytotoxic CD8+ TRM accumulation was due to chronic rejection and not transplantation alone. Furthermore, blocking NKG2D in vivo attenuated the airway remodeling following transplantation and diminished airway accumulation of CD8+ T cells. Our findings support NKG2D as a potential therapeutic target for CLAD, affecting cytotoxic CD8+ TRM accumulation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Tim Oury
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | | | - John F. McDyer
- Department of Medicine
- Starzl Transplantation Institute, Pittsburgh, Pennsylvania, USA
| | | | - Mark E. Snyder
- Department of Medicine
- Starzl Transplantation Institute, Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
3
|
Charmetant X, Pettigrew GJ, Thaunat O. Allorecognition Unveiled: Integrating Recent Breakthroughs Into the Current Paradigm. Transpl Int 2024; 37:13523. [PMID: 39588197 PMCID: PMC11586167 DOI: 10.3389/ti.2024.13523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 10/29/2024] [Indexed: 11/27/2024]
Abstract
In transplantation, genetic differences between donor and recipient trigger immune responses that cause graft rejection. Allorecognition, the process by which the immune system discriminates allogeneic grafts, targets major histocompatibility complex (MHC) and minor histocompatibility antigens. Historically, it was believed that allorecognition was solely mediated by the recipient's adaptive immune system recognizing donor-specific alloantigens. However, recent research has shown significant roles for innate immune components, such as lymphoid and myeloid cells, which are sometimes triggered by the mere absence of a self-protein in the graft. This review integrates recent breakthroughs into the current allorecognition paradigm based on the well-established direct and indirect pathways, emphasizing the semi-direct pathway where recipient antigen-presenting cells (APCs) acquire donor MHC molecules, and the inverted direct pathway where donor CD4+ T cells within the graft activate recipient B cells to produce donor-specific antibodies (DSAs). The review also explores the role of natural killer (NK) cells in both promoting and inhibiting graft rejection, highlighting their dual role in innate allorecognition. Additionally, it discusses the emerging understanding of myeloid cell-mediated allorecognition and its implications for initiating adaptive immune responses. These insights aim to provide a more comprehensive understanding of allorecognition, potentially leading to improved transplant outcomes.
Collapse
Affiliation(s)
- Xavier Charmetant
- Centre International de Recherche en Infectiologie, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, University Lyon, Lyon, France
- Department of Transplantation, Nephrology and Clinical Immunology, Edouard Herriot Hospital, Hospices Civils de Lyon, Lyon, France
- Lyon-Est Faculty of Medicine, Claude Bernard University (Lyon 1), Villeurbanne, France
| | - Gavin J. Pettigrew
- Department of Surgery, University of Cambridge, Cambridge, United Kingdom
| | - Olivier Thaunat
- Centre International de Recherche en Infectiologie, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, University Lyon, Lyon, France
- Department of Transplantation, Nephrology and Clinical Immunology, Edouard Herriot Hospital, Hospices Civils de Lyon, Lyon, France
- Lyon-Est Faculty of Medicine, Claude Bernard University (Lyon 1), Villeurbanne, France
| |
Collapse
|
4
|
Tsao T, Qiu L, Bharti R, Shemesh A, Hernandez AM, Cleary SJ, Greenland NY, Santos J, Shi R, Bai L, Richardson J, Dilley K, Will M, Tomasevic N, Sputova T, Salles A, Kang J, Zhang D, Hays SR, Kukreja J, Singer JP, Lanier LL, Looney MR, Greenland JR, Calabrese DR. CD94 + natural killer cells potentiate pulmonary ischaemia-reperfusion injury. Eur Respir J 2024; 64:2302171. [PMID: 39190789 DOI: 10.1183/13993003.02171-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 06/30/2024] [Indexed: 08/29/2024]
Abstract
BACKGROUND Pulmonary ischaemia-reperfusion injury (IRI) is a major contributor to poor lung transplant outcomes. We recently demonstrated a central role of airway-centred natural killer (NK) cells in mediating IRI; however, there are no existing effective therapies for directly targeting NK cells in humans. METHODS We hypothesised that a depleting anti-CD94 monoclonal antibody (mAb) would provide therapeutic benefit in mouse and human models of IRI based on high levels of KLRD1 (CD94) transcripts in bronchoalveolar lavage samples from lung transplant patients. RESULTS We found that CD94 is highly expressed on mouse and human NK cells, with increased expression during IRI. Anti-mouse and anti-human mAbs against CD94 showed effective NK cell depletion in mouse and human models and blunted lung damage and airway epithelial killing, respectively. In two different allogeneic orthotopic lung transplant mouse models, anti-CD94 treatment during induction reduced early lung injury and chronic inflammation relative to control therapies. Anti-CD94 did not increase donor antigen-presenting cells that could alter long-term graft acceptance. CONCLUSIONS Lung transplant induction regimens incorporating anti-CD94 treatment may safely improve early clinical outcomes.
Collapse
Affiliation(s)
- Tasha Tsao
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
- T. Tsao and L. Qiu contributed equally
| | - Longhui Qiu
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
- T. Tsao and L. Qiu contributed equally
| | - Reena Bharti
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Avishai Shemesh
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy San Francisco, San Francisco, CA, USA
| | - Alberto M Hernandez
- Parker Institute for Cancer Immunotherapy San Francisco, San Francisco, CA, USA
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, USA
| | - Simon J Cleary
- Institute of Pharmaceutical Science, King's College London, London, UK
| | - Nancy Y Greenland
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
| | - Jesse Santos
- Department of Surgery, University of California San Francisco - East Bay, Oakland, CA, USA
| | | | - Lu Bai
- Dren Bio, Foster City, CA, USA
| | | | | | | | | | | | | | | | - Dongliang Zhang
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Steven R Hays
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Jasleen Kukreja
- Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Jonathan P Singer
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Lewis L Lanier
- Parker Institute for Cancer Immunotherapy San Francisco, San Francisco, CA, USA
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, USA
| | - Mark R Looney
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - John R Greenland
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
- Medical Service, Veterans Affairs Health Care System, San Francisco, CA, USA
| | - Daniel R Calabrese
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
- Medical Service, Veterans Affairs Health Care System, San Francisco, CA, USA
| |
Collapse
|
5
|
Ashraf MI, Mengwasser J, Reutzel-Selke A, Polenz D, Führer K, Lippert S, Tang P, Michaelis E, Catar R, Pratschke J, Witzel C, Sauer IM, Tullius SG, Kern B. Depletion of donor dendritic cells ameliorates immunogenicity of both skin and hind limb transplants. Front Immunol 2024; 15:1395945. [PMID: 38799435 PMCID: PMC11116604 DOI: 10.3389/fimmu.2024.1395945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 04/15/2024] [Indexed: 05/29/2024] Open
Abstract
Acute cellular rejection remains a significant obstacle affecting successful outcomes of organ transplantation including vascularized composite tissue allografts (VCA). Donor antigen presenting cells (APCs), particularly dendritic cells (DCs), orchestrate early alloimmune responses by activating recipient effector T cells. Employing a targeted approach, we investigated the impact of donor-derived conventional DCs (cDCs) and APCs on the immunogenicity of skin and skin-containing VCA grafts, using mouse models of skin and hind limb transplantation. By post-transplantation day 6, skin grafts demonstrated severe rejections, characterized by predominance of recipient CD4 T cells. In contrast, hind limb grafts showed moderate rejection, primarily infiltrated by CD8 T cells. Notably, the skin component exhibited heightened immunogenicity when compared to the entire VCA, evidenced by increased frequencies of pan (CD11b-CD11c+), mature (CD11b-CD11c+MHCII+) and active (CD11b-CD11c+CD40+) DCs and cDC2 subset (CD11b+CD11c+ MHCII+) in the lymphoid tissues and the blood of skin transplant recipients. While donor depletion of cDC and APC reduced frequencies, maturation and activation of DCs in all analyzed tissues of skin transplant recipients, reduction in DC activities was only observed in the spleen of hind limb recipients. Donor cDC and APC depletion did not impact all lymphocyte compartments but significantly affected CD8 T cells and activated CD4 T in lymph nodes of skin recipients. Moreover, both donor APC and cDC depletion attenuated the Th17 immune response, evident by significantly reduced Th17 (CD4+IL-17+) cells in the spleen of skin recipients and reduced levels of IL-17E and lymphotoxin-α in the serum samples of both skin and hind limb recipients. In conclusion, our findings underscore the highly immunogenic nature of skin component in VCA. The depletion of donor APCs and cDCs mitigates the immunogenicity of skin grafts while exerting minimal impact on VCA.
Collapse
Affiliation(s)
- Muhammad Imtiaz Ashraf
- Department of Surgery, Experimental Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Joerg Mengwasser
- Department of Surgery, Experimental Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
- Department of General, Visceral and Transplant Surgery, Hannover Medical School, Hannover, Germany
| | - Anja Reutzel-Selke
- Department of Surgery, Experimental Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Dietrich Polenz
- Department of Surgery, Experimental Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Kirsten Führer
- Department of Surgery, Experimental Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Steffen Lippert
- Department of Surgery, Experimental Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Peter Tang
- Department of Surgery, Experimental Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Edward Michaelis
- Department of Pathology, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Rusan Catar
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Healthy, Berlin, Germany
| | - Johann Pratschke
- Department of Surgery, Experimental Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Christian Witzel
- Department of Plastic Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Igor M. Sauer
- Department of Surgery, Experimental Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Stefan G. Tullius
- Division of Transplant Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Einstein Berlin Institute of Health Visiting Fellow, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Barbara Kern
- Department of Surgery, Experimental Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
- Department of Plastic Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin Institute of Health (BIH) Biomedical Innovation Academy, Berlin Institute of Health (BIH) Charité Clinician Scientist Program, Berlin, Germany
| |
Collapse
|
6
|
Vidal-dos-Santos M, Anunciação LF, Armstrong-Jr R, Ricardo-da-Silva FY, Ramos IYT, Correia CJ, Moreira LFP, Leuvenink HGD, Breithaupt-Faloppa AC. 17β-estradiol and methylprednisolone association as a therapeutic option to modulate lung inflammation in brain-dead female rats. Front Immunol 2024; 15:1375943. [PMID: 38765005 PMCID: PMC11099279 DOI: 10.3389/fimmu.2024.1375943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 04/15/2024] [Indexed: 05/21/2024] Open
Abstract
Introduction Brain death (BD) is known to compromise graft quality by causing hemodynamic, metabolic, and hormonal changes. The abrupt reduction of female sex hormones after BD was associated with increased lung inflammation. The use of both corticoids and estradiol independently has presented positive results in modulating BD-induced inflammatory response. However, studies have shown that for females the presence of both estrogen and corticoids is necessary to ensure adequate immune response. In that sense, this study aims to investigate how the association of methylprednisolone (MP) and estradiol (E2) could modulate the lung inflammation triggered by BD in female rats. Methods Female Wistar rats (8 weeks) were divided into four groups: sham (animals submitted to the surgical process, without induction of BD), BD (animals submitted to BD), MP/E2 (animals submitted to BD that received MP and E2 treatment 3h after BD induction) and MP (animals submitted to BD that received MP treatment 3h after BD induction). Results Hemodynamics, systemic and local quantification of IL-6, IL-1β, VEGF, and TNF-α, leukocyte infiltration to the lung parenchyma and airways, and adhesion molecule expression were analyzed. After treatment, MP/E2 association was able to reinstate mean arterial pressure to levels close to Sham animals (p<0.05). BD increased leukocyte infiltration to the airways and MP/E2 was able to reduce the number of cells (p=0.0139). Also, the associated treatment modulated the vasculature by reducing the expression of VEGF (p=0.0616) and maintaining eNOS levels (p=0.004) in lung tissue. Discussion Data presented in this study show that the association between corticoids and estradiol could represent a better treatment strategy for lung inflammation in the female BD donor by presenting a positive effect in the hemodynamic management of the donor, as well as by reducing infiltrated leukocyte to the airways and release of inflammatory markers in the short and long term.
Collapse
Affiliation(s)
- Marina Vidal-dos-Santos
- Laboratório de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- Department of Surgery, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | - Lucas F. Anunciação
- Laboratório de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Roberto Armstrong-Jr
- Laboratório de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Fernanda Y. Ricardo-da-Silva
- Laboratório de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Isabella Yumi Taira Ramos
- Laboratório de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Cristiano J. Correia
- Laboratório de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Luiz F. P. Moreira
- Laboratório de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Henri G. D. Leuvenink
- Department of Surgery, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | - Ana C. Breithaupt-Faloppa
- Laboratório de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
7
|
Santos J, Wang P, Shemesh A, Liu F, Tsao T, Aguilar OA, Cleary SJ, Singer JP, Gao Y, Hays SR, Golden JA, Leard L, Kleinhenz ME, Kolaitis NA, Shah R, Venado A, Kukreja J, Weigt SS, Belperio JA, Lanier LL, Looney MR, Greenland JR, Calabrese DR. CCR5 drives NK cell-associated airway damage in pulmonary ischemia-reperfusion injury. JCI Insight 2023; 8:e173716. [PMID: 37788115 PMCID: PMC10721259 DOI: 10.1172/jci.insight.173716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 09/20/2023] [Indexed: 10/05/2023] Open
Abstract
Primary graft dysfunction (PGD) limits clinical benefit after lung transplantation, a life-prolonging therapy for patients with end-stage disease. PGD is the clinical syndrome resulting from pulmonary ischemia-reperfusion injury (IRI), driven by innate immune inflammation. We recently demonstrated a key role for NK cells in the airways of mouse models and human tissue samples of IRI. Here, we used 2 mouse models paired with human lung transplant samples to investigate the mechanisms whereby NK cells migrate to the airways to mediate lung injury. We demonstrate that chemokine receptor ligand transcripts and proteins are increased in mouse and human disease. CCR5 ligand transcripts were correlated with NK cell gene signatures independently of NK cell CCR5 ligand secretion. NK cells expressing CCR5 were increased in the lung and airways during IRI and had increased markers of tissue residency and maturation. Allosteric CCR5 drug blockade reduced the migration of NK cells to the site of injury. CCR5 blockade also blunted quantitative measures of experimental IRI. Additionally, in human lung transplant bronchoalveolar lavage samples, we found that CCR5 ligand was associated with increased patient morbidity and that the CCR5 receptor was increased in expression on human NK cells following PGD. These data support a potential mechanism for NK cell migration during lung injury and identify a plausible preventative treatment for PGD.
Collapse
Affiliation(s)
- Jesse Santos
- Department of Medicine, UCSF, San Francisco, California, USA
- Department of Surgery, UCSF - East Bay, Oakland, California, USA
| | - Ping Wang
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Avishai Shemesh
- Department of Medicine, UCSF, San Francisco, California, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
| | - Fengchun Liu
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Tasha Tsao
- Department of Medicine, UCSF, San Francisco, California, USA
| | | | - Simon J. Cleary
- Department of Medicine, UCSF, San Francisco, California, USA
| | | | - Ying Gao
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Steven R. Hays
- Department of Medicine, UCSF, San Francisco, California, USA
| | | | - Lorriana Leard
- Department of Medicine, UCSF, San Francisco, California, USA
| | | | | | - Rupal Shah
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Aida Venado
- Department of Medicine, UCSF, San Francisco, California, USA
| | | | - S. Sam Weigt
- Department of Medicine, UCLA, Los Angeles, California, USA
| | | | - Lewis L. Lanier
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
- Department of Microbiology and Immunology, and
| | - Mark R. Looney
- Department of Medicine, UCSF, San Francisco, California, USA
| | - John R. Greenland
- Department of Medicine, UCSF, San Francisco, California, USA
- Medical Service, Veterans Affairs Health Care System, San Francisco, California, USA
| | - Daniel R. Calabrese
- Department of Medicine, UCSF, San Francisco, California, USA
- Medical Service, Veterans Affairs Health Care System, San Francisco, California, USA
| |
Collapse
|
8
|
Mak ML, Reid KT, Crome SQ. Protective and pathogenic functions of innate lymphoid cells in transplantation. Clin Exp Immunol 2023; 213:23-39. [PMID: 37119279 PMCID: PMC10324558 DOI: 10.1093/cei/uxad050] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/27/2023] [Accepted: 04/28/2023] [Indexed: 05/01/2023] Open
Abstract
Innate lymphoid cells (ILCs) are a family of lymphocytes with essential roles in tissue homeostasis and immunity. Along with other tissue-resident immune populations, distinct subsets of ILCs have important roles in either promoting or inhibiting immune tolerance in a variety of contexts, including cancer and autoimmunity. In solid organ and hematopoietic stem cell transplantation, both donor and recipient-derived ILCs could contribute to immune tolerance or rejection, yet understanding of protective or pathogenic functions are only beginning to emerge. In addition to roles in directing or regulating immune responses, ILCs interface with parenchymal cells to support tissue homeostasis and even regeneration. Whether specific ILCs are tissue-protective or enhance ischemia reperfusion injury or fibrosis is of particular interest to the field of transplantation, beyond any roles in limiting or promoting allograft rejection or graft-versus host disease. Within this review, we discuss the current understanding of ILCs functions in promoting immune tolerance and tissue repair at homeostasis and in the context of transplantation and highlight where targeting or harnessing ILCs could have applications in novel transplant therapies.
Collapse
Affiliation(s)
- Martin L Mak
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
- Toronto General Hospital Research Institute, Ajmera Transplant Centre, University Health Network, Toronto, Canada
| | - Kyle T Reid
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
- Toronto General Hospital Research Institute, Ajmera Transplant Centre, University Health Network, Toronto, Canada
| | - Sarah Q Crome
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
- Toronto General Hospital Research Institute, Ajmera Transplant Centre, University Health Network, Toronto, Canada
| |
Collapse
|
9
|
Liao M, Wang C, Zhang M, Qiao K. Insight on immune cells in rejection and infection postlung transplant. Immun Inflamm Dis 2023; 11:e868. [PMID: 37506156 PMCID: PMC10336664 DOI: 10.1002/iid3.868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 04/25/2023] [Accepted: 04/27/2023] [Indexed: 07/30/2023] Open
Abstract
OBJECTIVE The aim of this study is to provide a concise overview of the role of immune cells in rejection and infection after lung transplantation. METHODS Based on previous clinical and basic studies, the role of various types of immune cells in the development of rejection and infection after lung transplantation is summarized. RESULTS Immune cell functional status is strongly associated with common complications after lung transplantation, such as primary graft dysfunction, infection and occlusive bronchitis syndrome. Targeted balancing of immune cell tolerance and rejection is an important tool for successful lung transplantation. CONCLUSION A comprehensive understanding of immune cell function and the mechanisms that balance immune tolerance and immune rejection may be a crucial factor in improving survival after lung transplantation.
Collapse
Affiliation(s)
- Mingfeng Liao
- Guangdong Key Lab for Diagnosis & Treatment of Emerging Infectious DiseasesShenzhen Third People's HospitalShenzhenGuangdong ProvincePeople's Republic of China
| | - Chaoxi Wang
- Department of Thoracic SurgeryShenzhen Third People's HospitalShenzhenGuangdong ProvincePeople's Republic of China
| | - Mingxia Zhang
- Guangdong Key Lab for Diagnosis & Treatment of Emerging Infectious DiseasesShenzhen Third People's HospitalShenzhenGuangdong ProvincePeople's Republic of China
| | - Kun Qiao
- Department of Thoracic SurgeryShenzhen Third People's HospitalShenzhenGuangdong ProvincePeople's Republic of China
| |
Collapse
|
10
|
Biased IL-2 signals induce Foxp3-rich pulmonary lymphoid structures and facilitate long-term lung allograft acceptance in mice. Nat Commun 2023; 14:1383. [PMID: 36914624 PMCID: PMC10011523 DOI: 10.1038/s41467-023-36924-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 02/24/2023] [Indexed: 03/16/2023] Open
Abstract
Transplantation of solid organs can be life-saving in patients with end-stage organ failure, however, graft rejection remains a major challenge. In this study, by pre-conditioning with interleukin-2 (IL-2)/anti-IL-2 antibody complex treatment biased toward IL-2 receptor α, we achieved acceptance of fully mismatched orthotopic lung allografts that remained morphologically and functionally intact for more than 90 days in immunocompetent mice. These allografts are tolerated by the actions of forkhead box p3 (Foxp3)+ regulatory T (Treg) cells that home to the lung allografts. Although counts of circulating Treg cells rapidly return to baseline following cessation of IL-2 treatment, Foxp3+ Treg cells persist in peribronchial and peribronchiolar areas of the grafted lungs, forming organized clusters reminiscent of inducible tertiary lymphoid structures (iTLS). These iTLS in lung allografts are made of Foxp3+ Treg cells, conventional T cells, and B cells, as evidenced by using microscopy-based distribution and neighborhood analyses. Foxp3-transgenic mice with inducible and selective deletion of Foxp3+ cells are unable to form iTLS in lung allografts, and these mice acutely reject lung allografts. Collectively, we report that short-term, high-intensity and biased IL-2 pre-conditioning facilitates acceptance of vascularized and ventilated lung allografts without the need of immunosuppression, by inducing Foxp3-controlled iTLS formation within allografts.
Collapse
|
11
|
Watanabe T, Lam C, Oliver J, Oishi H, Teskey G, Beber S, Boonstra K, Mauricio Umaña J, Buhari H, Joe B, Guan Z, Horie M, Keshavjee S, Martinu T, Juvet SC. Donor Batf3 inhibits murine lung allograft rejection and airway fibrosis. Mucosal Immunol 2023; 16:104-120. [PMID: 36842540 DOI: 10.1016/j.mucimm.2023.02.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 02/14/2023] [Indexed: 02/28/2023]
Abstract
Chronic lung allograft dysfunction (CLAD) limits survival after lung transplantation. Noxious stimuli entering the airways foster CLAD development. Classical dendritic cells (cDCs) link innate and adaptive immunity and exhibit regional and functional specialization in the lung. The transcription factor basic leucine zipper ATF-like 3 (BATF3) is absolutely required for the development of type 1 cDCs (cDC1s), which reside in the airway epithelium and have variable responses depending on the context. We studied the role of BATF3 in a mouse minor alloantigen-mismatched orthotopic lung transplant model of CLAD with and without airway inflammation triggered by repeated administration of intratracheal lipopolysaccharide (LPS). We found that cDC1s accumulated in allografts compared with isografts and that donor cDC1s were gradually replaced by recipient cDC1s. LPS administration increased the number of cDC1s and enhanced their state of activation. We found that Batf3-/- recipient mice experienced reduced acute rejection in response to LPS; in contrast, Batf3-/- donor grafts underwent enhanced lung and skin allograft rejection and drove augmented recipient cluster of differentiation 8+ T-cell expansion in the absence of LPS. Our findings suggest that donor and recipient cDC1s have differing and context-dependent roles and may represent a therapeutic target in lung transplantation.
Collapse
Affiliation(s)
- Tatsuaki Watanabe
- Latner Thoracic Research Laboratories, University Health Network, University of Toronto, Toronto, Canada; Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Christina Lam
- Latner Thoracic Research Laboratories, University Health Network, University of Toronto, Toronto, Canada
| | - Jillian Oliver
- Latner Thoracic Research Laboratories, University Health Network, University of Toronto, Toronto, Canada
| | - Hisashi Oishi
- Latner Thoracic Research Laboratories, University Health Network, University of Toronto, Toronto, Canada; Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Grace Teskey
- Latner Thoracic Research Laboratories, University Health Network, University of Toronto, Toronto, Canada
| | - Samuel Beber
- Latner Thoracic Research Laboratories, University Health Network, University of Toronto, Toronto, Canada
| | - Kristen Boonstra
- Latner Thoracic Research Laboratories, University Health Network, University of Toronto, Toronto, Canada
| | - Juan Mauricio Umaña
- Latner Thoracic Research Laboratories, University Health Network, University of Toronto, Toronto, Canada
| | - Hifza Buhari
- Latner Thoracic Research Laboratories, University Health Network, University of Toronto, Toronto, Canada
| | - Betty Joe
- Latner Thoracic Research Laboratories, University Health Network, University of Toronto, Toronto, Canada
| | - Zehong Guan
- Latner Thoracic Research Laboratories, University Health Network, University of Toronto, Toronto, Canada
| | - Miho Horie
- Joint Department of Medical Imaging, University Health Network, Toronto, Canada
| | - Shaf Keshavjee
- Latner Thoracic Research Laboratories, University Health Network, University of Toronto, Toronto, Canada
| | - Tereza Martinu
- Latner Thoracic Research Laboratories, University Health Network, University of Toronto, Toronto, Canada
| | - Stephen C Juvet
- Latner Thoracic Research Laboratories, University Health Network, University of Toronto, Toronto, Canada.
| |
Collapse
|
12
|
Bos S, Milross L, Filby AJ, Vos R, Fisher AJ. Immune processes in the pathogenesis of chronic lung allograft dysfunction: identifying the missing pieces of the puzzle. Eur Respir Rev 2022; 31:31/165/220060. [PMID: 35896274 DOI: 10.1183/16000617.0060-2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 05/19/2022] [Indexed: 11/05/2022] Open
Abstract
Lung transplantation is the optimal treatment for selected patients with end-stage chronic lung diseases. However, chronic lung allograft dysfunction remains the leading obstacle to improved long-term outcomes. Traditionally, lung allograft rejection has been considered primarily as a manifestation of cellular immune responses. However, in reality, an array of complex, interacting and multifactorial mechanisms contribute to its emergence. Alloimmune-dependent mechanisms, including T-cell-mediated rejection and antibody-mediated rejection, as well as non-alloimmune injuries, have been implicated. Moreover, a role has emerged for autoimmune responses to lung self-antigens in the development of chronic graft injury. The aim of this review is to summarise the immune processes involved in the pathogenesis of chronic lung allograft dysfunction, with advanced insights into the role of innate immune pathways and crosstalk between innate and adaptive immunity, and to identify gaps in current knowledge.
Collapse
Affiliation(s)
- Saskia Bos
- Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK.,Institute of Transplantation, Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, UK
| | - Luke Milross
- Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK
| | - Andrew J Filby
- Flow Cytometry Core and Innovation, Methodology and Application Research Theme, Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Robin Vos
- Dept of CHROMETA, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium.,University Hospitals Leuven, Dept of Respiratory Diseases, Leuven, Belgium
| | - Andrew J Fisher
- Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK .,Institute of Transplantation, Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, UK
| |
Collapse
|
13
|
Santos J, Calabrese DR, Greenland JR. Lymphocytic Airway Inflammation in Lung Allografts. Front Immunol 2022; 13:908693. [PMID: 35911676 PMCID: PMC9335886 DOI: 10.3389/fimmu.2022.908693] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/16/2022] [Indexed: 11/16/2022] Open
Abstract
Lung transplant remains a key therapeutic option for patients with end stage lung disease but short- and long-term survival lag other solid organ transplants. Early ischemia-reperfusion injury in the form of primary graft dysfunction (PGD) and acute cellular rejection are risk factors for chronic lung allograft dysfunction (CLAD), a syndrome of airway and parenchymal fibrosis that is the major barrier to long term survival. An increasing body of research suggests lymphocytic airway inflammation plays a significant role in these important clinical syndromes. Cytotoxic T cells are observed in airway rejection, and transcriptional analysis of airways reveal common cytotoxic gene patterns across solid organ transplant rejection. Natural killer (NK) cells have also been implicated in the early allograft damage response to PGD, acute rejection, cytomegalovirus, and CLAD. This review will examine the roles of lymphocytic airway inflammation across the lifespan of the allograft, including: 1) The contribution of innate lymphocytes to PGD and the impact of PGD on the adaptive immune response. 2) Acute cellular rejection pathologies and the limitations in identifying airway inflammation by transbronchial biopsy. 3) Potentiators of airway inflammation and heterologous immunity, such as respiratory infections, aspiration, and the airway microbiome. 4) Airway contributions to CLAD pathogenesis, including epithelial to mesenchymal transition (EMT), club cell loss, and the evolution from constrictive bronchiolitis to parenchymal fibrosis. 5) Protective mechanisms of fibrosis involving regulatory T cells. In summary, this review will examine our current understanding of the complex interplay between the transplanted airway epithelium, lymphocytic airway infiltration, and rejection pathologies.
Collapse
Affiliation(s)
- Jesse Santos
- Department of Medicine University of California, San Francisco, San Francisco, CA, United States
| | - Daniel R. Calabrese
- Department of Medicine University of California, San Francisco, San Francisco, CA, United States
- Medical Service, Veterans Affairs Health Care System, San Francisco, CA, United States
| | - John R. Greenland
- Department of Medicine University of California, San Francisco, San Francisco, CA, United States
- Medical Service, Veterans Affairs Health Care System, San Francisco, CA, United States
| |
Collapse
|
14
|
Zamir MR, Shahi A, Salehi S, Amirzargar A. Natural killer cells and killer cell immunoglobulin-like receptors in solid organ transplantation: Protectors or opponents? Transplant Rev (Orlando) 2022; 36:100723. [DOI: 10.1016/j.trre.2022.100723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/20/2022] [Accepted: 07/25/2022] [Indexed: 10/16/2022]
|
15
|
Bai YZ, Roberts SH, Kreisel D, Nava RG. Microbiota in heart and lung transplantation: implications for innate-adaptive immune interface. Curr Opin Organ Transplant 2021; 26:609-614. [PMID: 34561360 DOI: 10.1097/mot.0000000000000923] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW Transplantation continues to be the only treatment option for end-stage organ failure when other interventions have failed. Although short-term outcomes have improved due to advances in perioperative care, long-term outcomes continue to be adversely affected by chronic rejection. Little is known about the role microbiota play in modulating alloimmune responses and potentially contributing to graft failure. Initial data have identified a correlation between specific changes of the recipient and/or donor microbiota and transplant outcomes. In this review, we will focus on recent findings concerning the complex interplay between microbiota and the innate immune system after heart and lung transplantation. RECENT FINDINGS Gut microbiome derangements in heart failure promote an inflammatory state and have lasting effects on the innate immune system, with an observed association between increased levels of microbiota-dependent metabolites and acute rejection after cardiac transplantation. The lung allograft microbiome interacts with components of the innate immune system, such as toll-like receptor signalling pathways, NKG2C+ natural killer cells and the NLRP3 inflammasome, to alter posttransplant outcomes, which may result in the development of chronic rejection. SUMMARY The innate immune system is influenced by alterations in the microbiome before and after heart and lung transplantation, thereby offering potential therapeutic targets for prolonging allograft survival.
Collapse
Affiliation(s)
| | | | - Daniel Kreisel
- Department of Surgery
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | | |
Collapse
|
16
|
Lin CM, Gill RG, Mehrad B. The natural killer cell activating receptor, NKG2D, is critical to antibody-dependent chronic rejection in heart transplantation. Am J Transplant 2021; 21:3550-3560. [PMID: 34014614 PMCID: PMC9036609 DOI: 10.1111/ajt.16690] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 04/26/2021] [Accepted: 05/04/2021] [Indexed: 01/25/2023]
Abstract
Chronic rejection is among the most pressing clinical challenges in solid organ transplantation. Interestingly, in a mouse model of heterotopic heart transplantation, antibody-dependent, natural killer (NK) cell-mediated chronic cardiac allograft vasculopathy occurs in some donor-recipient strain combinations, but not others. In this study, we sought to identify the mechanism underlying this unexplained phenomenon. Cardiac allografts from major histocompatibility complex (MHC) mismatched donors were transplanted into immune-deficient C57Bl/6.rag-/- recipients, followed by administration of a monoclonal antibody against the donor MHC class I antigen. We found marked allograft vasculopathy in hearts from C3H donors, but near-complete protection of BALB/c allografts from injury. We found no difference in recipient NK cell phenotype or intrinsic responsiveness to activating signals between recipients of C3H versus BALB/c allografts. However, cardiac endothelial cells from C3H allografts showed an approximately twofold higher expression of Rae-1, an activating ligand of the NK cell receptor natural killer group 2D (NKG2D). Importantly, the administration of a neutralizing antibody against NKG2D abrogated the development of allograft vasculopathy in recipients of C3H allografts, even in the presence of donor-specific antibodies. Therefore, the activating NK cell receptor NKG2D is necessary in this model of chronic cardiac allograft vasculopathy, and strain-dependent expression of NK activating ligands correlates with the development of this disease.
Collapse
Affiliation(s)
- Christine M. Lin
- University of Florida. Department of Medicine (Gainesville, FL, USA)
| | - Ronald G. Gill
- University of Colorado, Anschutz Medical Campus. Department of Surgery (Aurora, CO, USA)
| | - Borna Mehrad
- University of Florida. Department of Medicine (Gainesville, FL, USA)
| |
Collapse
|
17
|
Shepherd HM, Gauthier JM, Kreisel D. Tolerance, immunosuppression, and immune modulation: impacts on lung allograft survival. Curr Opin Organ Transplant 2021; 26:328-332. [PMID: 33782247 PMCID: PMC8523032 DOI: 10.1097/mot.0000000000000871] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE OF REVIEW Immune responses following lung transplantation continue to result in high rates of allograft failure and rejection, and current immunosuppression does not address the unique immunologic properties of the lung. Here, we review recent studies on lung allograft tolerance and alloimmunity and discuss implications for immunosuppression. RECENT FINDINGS Processes governing tolerance and alloimmunity in lung allografts differ from other solid organs. Recent studies have suggested that allorecognition is regulated at the level of the lung graft. Furthermore, certain cell populations essential for lung allograft tolerance may facilitate rejection in other organs. Induction of lung allograft tolerance is associated with the formation of tertiary lymphoid organs, which are enriched in regulatory T cells and play an important role in preventing rejection. SUMMARY Recent discoveries regarding alloactivation and the regulation of tolerance following lung transplantation have introduced exciting potential avenues for the development of lung-specific immunosuppression.
Collapse
Affiliation(s)
- Hailey M. Shepherd
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO
| | - Jason M. Gauthier
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO
| | - Daniel Kreisel
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO
| |
Collapse
|
18
|
Hennessy C, Lewik G, Cross A, Hester J, Issa F. Recent advances in our understanding of the allograft response. Fac Rev 2021; 10:21. [PMID: 33718938 PMCID: PMC7946390 DOI: 10.12703/r/10-21] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Organ transplantation is a life-saving treatment for end-stage organ failure. However, despite advances in immunosuppression, donor matching, tissue typing, and organ preservation, many organs are still lost each year to rejection. Ultimately, tolerance in the absence of immunosuppression is the goal, and although this seldom occurs spontaneously, a deeper understanding of alloimmunity may provide avenues for future therapies which aid in its establishment. Here, we highlight the recent key advances in our understanding of the allograft response. On the innate side, recent work has highlighted the previously unrecognised role of innate lymphoid cells as well as natural killer cells in promoting the alloresponse. The two major routes of allorecognition have recently been joined by a third newly identified pathway, semi-direct allorecognition, which is proving to be a key active pathway in transplantation. Through this review, we detail these newly defined areas in the allograft response and highlight areas for potential future therapeutic intervention.
Collapse
Affiliation(s)
- Conor Hennessy
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Guido Lewik
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Amy Cross
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Joanna Hester
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Fadi Issa
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| |
Collapse
|
19
|
Tanaka S, Gauthier JM, Terada Y, Takahashi T, Li W, Hashimoto K, Higashikubo R, Hachem RR, Bharat A, Ritter JH, Nava RG, Puri V, Krupnick AS, Gelman AE, Kreisel D. Bacterial products in donor airways prevent the induction of lung transplant tolerance. Am J Transplant 2021; 21:353-361. [PMID: 32786174 PMCID: PMC7775268 DOI: 10.1111/ajt.16256] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 07/17/2020] [Accepted: 08/03/2020] [Indexed: 01/25/2023]
Abstract
Although postoperative bacterial infections can trigger rejection of pulmonary allografts, the impact of bacterial colonization of donor grafts on alloimmune responses to transplanted lungs remains unknown. Here, we tested the hypothesis that bacterial products present within donor grafts at the time of implantation promote lung allograft rejection. Administration of the toll-like receptor 2 (TLR2) agonist Pam3 Cys4 to Balb/c wild-type grafts triggered acute cellular rejection after transplantation into B6 wild-type recipients that received perioperative costimulatory blockade. Pam3 Cys4 -triggered rejection was associated with an expansion of CD8+ T lymphocytes and CD11c+ CD11bhi MHC (major histocompatibility complex) class II+ antigen-presenting cells within the transplanted lungs. Rejection was prevented when lungs were transplanted into TLR2-deficient recipients but not when MyD88-deficient donors were used. Adoptive transfer of B6 wild-type monocytes, but not T cells, following transplantation into B6 TLR2-deficient recipients restored the ability of Pam3 Cys4 to trigger acute cellular rejection. Thus, we have demonstrated that activation of TLR2 by a bacterial lipopeptide within the donor airways prevents the induction of lung allograft tolerance through a process mediated by recipient-derived monocytes. Our work suggests that donor lungs harboring bacteria may precipitate an inflammatory response that can facilitate allograft rejection.
Collapse
Affiliation(s)
- Satona Tanaka
- Department of Surgery, Washington University, Saint Louis, MO
| | | | - Yuriko Terada
- Department of Surgery, Washington University, Saint Louis, MO
| | | | - Wenjun Li
- Department of Surgery, Washington University, Saint Louis, MO
| | - Kohei Hashimoto
- Department of Surgery, Washington University, Saint Louis, MO
| | | | | | - Ankit Bharat
- Department of Surgery, Northwestern University, Chicago, IL
| | - Jon H. Ritter
- Department of Pathology & Immunology, Washington University, Saint Louis, MO
| | - Ruben G. Nava
- Department of Surgery, Washington University, Saint Louis, MO
| | - Varun Puri
- Department of Surgery, Washington University, Saint Louis, MO
| | | | - Andrew E. Gelman
- Department of Surgery, Washington University, Saint Louis, MO,Department of Pathology & Immunology, Washington University, Saint Louis, MO
| | - Daniel Kreisel
- Department of Surgery, Washington University, Saint Louis, MO,Department of Pathology & Immunology, Washington University, Saint Louis, MO
| |
Collapse
|
20
|
Xiu MX, Liu YM, Wang WJ. Investigation of hub genes and immune status in heart transplant rejection using endomyocardial biopsies. J Cell Mol Med 2020; 25:763-773. [PMID: 33230903 PMCID: PMC7812257 DOI: 10.1111/jcmm.16127] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/26/2020] [Accepted: 11/06/2020] [Indexed: 12/13/2022] Open
Abstract
T cell‒mediated rejection (TCMR) and antibody‐mediated rejection (ABMR) are severe post‐transplantation complications for heart transplantation (HTx), whose molecular and immunological pathogenesis remains unclear. In the present study, the mRNA microarray data set GSE124897 containing 645 stable, 52 TCMR and 144 ABMR endomyocardial biopsies was obtained to screen for differentially expressed genes (DEGs) between rejected and stable HTx samples and to investigate immune cell infiltration. Functional enrichment analyses indicated roles of the DEGs primarily in immune‐related mechanisms. Protein‐protein interaction networks were then constructed, and ICAM1, CD44, HLA‐A and HLA‐B were identified as hub genes using the maximal clique centrality method. Immune cell infiltration analysis revealed differences in adaptive and innate immune cell populations between TCMR, ABMR and stable HTx samples. Additionally, hub gene expression levels significantly correlated with the degree and composition of immune cell infiltration in HTx rejection samples. Furthermore, drug‐gene interactions were constructed, and 12 FDA‐approved drugs were predicted to target hub genes. Finally, an external GSE2596 data set was used to validate the expression of the hub genes, and ROC curves indicated all four hub genes had promising diagnostic value for HTx rejection. This study provides a comprehensive perspective of molecular and immunological regulatory mechanisms underlying HTx rejection.
Collapse
Affiliation(s)
- Meng-Xi Xiu
- Medical School of Nanchang University, Nanchang, China
| | - Yuan-Meng Liu
- Medical School of Nanchang University, Nanchang, China
| | - Wen-Jun Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
21
|
Xiu MX, Liu ZT, Tang J. Screening and identification of key regulatory connections and immune cell infiltration characteristics for lung transplant rejection using mucosal biopsies. Int Immunopharmacol 2020; 87:106827. [PMID: 32791489 PMCID: PMC7417178 DOI: 10.1016/j.intimp.2020.106827] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 07/03/2020] [Accepted: 07/20/2020] [Indexed: 02/06/2023]
Abstract
This study aimed to explore key regulatory connections underlying lung transplant rejection. The differentially expressed genes (DEGs) between rejection and stable lung transplantation (LTx) samples were screened using R package limma, followed by functional enrichment analysis and protein-protein interaction network construction. Subsequently, a global triple network, including miRNAs, mRNAs, and transcription factors (TFs), was constructed. Furthermore, immune cell infiltration characteristics were analyzed to investigate the molecular immunology of lung transplant rejection. Finally, potential drug-target interactions were generated. In brief, 739 DEGs were found between rejection and stable LTx samples. PTPRC, IL-6, ITGAM, CD86, TLR8, TYROBP, CXCL10, ITGB2, and CCR5 were defined as hub genes. Eight TFs, including STAT1, SPIB, NFKB1, SPI1, STAT5A, RUNX1, VENTX, and BATF, and five miRNAs, including miR-335-5p, miR-26b-5p, miR-124-3p, miR-1-3p, and miR-155-5p, were involved in regulating hub genes. The immune cell infiltration analysis revealed higher proportions of activated memory CD4 T cells, follicular helper T cells, γδ T cells, monocytes, M1 and M2 macrophages, and eosinophils in rejection samples, besides lower proportions of resting memory CD4 T cells, regulatory T cells, activated NK cells, M0 macrophages, and resting mast cells. This study provided a comprehensive perspective of the molecular co-regulatory network underlying lung transplant rejection.
Collapse
Affiliation(s)
- Meng-Xi Xiu
- Medical School of Nanchang University, Nanchang, PR China
| | - Zu-Ting Liu
- Medical School of Nanchang University, Nanchang, PR China
| | - Jian Tang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, PR China.
| |
Collapse
|
22
|
Frye CC, Bery AI, Kreisel D, Kulkarni HS. Sterile inflammation in thoracic transplantation. Cell Mol Life Sci 2020; 78:581-601. [PMID: 32803398 DOI: 10.1007/s00018-020-03615-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 07/20/2020] [Accepted: 08/07/2020] [Indexed: 02/08/2023]
Abstract
The life-saving benefits of organ transplantation can be thwarted by allograft dysfunction due to both infectious and sterile inflammation post-surgery. Sterile inflammation can occur after necrotic cell death due to the release of endogenous ligands [such as damage-associated molecular patterns (DAMPs) and alarmins], which perpetuate inflammation and ongoing cellular injury via various signaling cascades. Ischemia-reperfusion injury (IRI) is a significant contributor to sterile inflammation after organ transplantation and is associated with detrimental short- and long-term outcomes. While the vicious cycle of sterile inflammation and cellular injury is remarkably consistent amongst different organs and even species, we have begun understanding its mechanistic basis only over the last few decades. This understanding has resulted in the developments of novel, yet non-specific therapies for mitigating IRI-induced graft damage, albeit with moderate results. Thus, further understanding of the mechanisms underlying sterile inflammation after transplantation is critical for identifying personalized therapies to prevent or interrupt this vicious cycle and mitigating allograft dysfunction. In this review, we identify common and distinct pathways of post-transplant sterile inflammation across both heart and lung transplantation that can potentially be targeted.
Collapse
Affiliation(s)
- C Corbin Frye
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| | - Amit I Bery
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, 4523 Clayton Avenue, Campus Box 8052, St. Louis, MO, 63110, USA.
| | - Daniel Kreisel
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Hrishikesh S Kulkarni
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, 4523 Clayton Avenue, Campus Box 8052, St. Louis, MO, 63110, USA
| |
Collapse
|
23
|
Critchley WR, Stone JP, Liao Q, Qin G, Risnes I, Trafford A, Scott H, Sjöberg T, Steen S, Fildes JE. Non-ischemic Heart Preservation via Hypothermic Cardioplegic Perfusion Induces Immunodepletion of Donor Hearts Resulting in Diminished Graft Infiltration Following Transplantation. Front Immunol 2020; 11:1621. [PMID: 32849549 PMCID: PMC7399062 DOI: 10.3389/fimmu.2020.01621] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 06/17/2020] [Indexed: 12/17/2022] Open
Abstract
Introduction: Many donor organs contain significant leukocyte reservoirs which upon transplantation activate recipient leukocytes to initiate acute rejection. We aimed to assess whether non-ischemic heart preservation via ex vivo perfusion promotes immunodepletion and alters the inflammatory status of the donor organ prior to transplantation. Methods: Isolated porcine hearts underwent ex vivo hypothermic, cardioplegic perfusion for 8 h. Leukocyte populations were quantified in left ventricle samples by flow cytometry. Cell-free DNA, cytokines, and chemokines were quantified in the perfusate. Tissue integrity was profiled by targeted proteomics and a histological assessment was performed. Heterotopic transplants comparing ex vivo hypothermic preservation and static cold storage were utilized to assess graft infiltration as a solid clinical endpoint. Results: Ex vivo perfusion significantly immunodepleted myocardial tissue. The perfusate displayed a selective, pro-inflammatory cytokine/chemokine pattern dominated by IFN-γ. The tissue molecular profile was improved following perfusion by diminished expression of nine pro-apoptotic and six ischemia-associated proteins. Histologically, no evidence of tissue damage was observed and cardiac troponin I was low throughout perfusion. Cell-free DNA was detected, the source of which may be necrotic/apoptotic leukocytes. Post-transplant graft infiltration was markedly reduced in terms of both leucocyte distribution and intensity of foci. Conclusions: These findings demonstrate that ex vivo perfusion significantly reduced donor heart immunogenicity via loss of resident leukocytes. Despite the pro-inflammatory cytokine pattern observed, a pro-survival and reduced ischemia-related profile was observed, indicating an improvement in graft viability by perfusion. Diminished graft infiltration was observed in perfused hearts compared with those preserved by static cold storage following 48 h of transplantation.
Collapse
Affiliation(s)
- William R Critchley
- The Ex-Vivo Lab, Division of Cell Matrix and Regenerative Medicine, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, School of Biological Sciences, The University of Manchester, Manchester, United Kingdom.,The Transplant Centre, Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom
| | - John P Stone
- The Ex-Vivo Lab, Division of Cell Matrix and Regenerative Medicine, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, School of Biological Sciences, The University of Manchester, Manchester, United Kingdom.,The Transplant Centre, Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom
| | - Qiuming Liao
- Department of Cardiothoracic Surgery, Lund University and Skåne University Hospital, Lund, Sweden
| | - Guangqi Qin
- Department of Cardiothoracic Surgery, Lund University and Skåne University Hospital, Lund, Sweden
| | - Ivar Risnes
- Department of Thoracic Surgery, Rikshospitalet, Oslo, Norway
| | - Andrew Trafford
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom
| | - Helge Scott
- Department of Pathology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Trygve Sjöberg
- Department of Cardiothoracic Surgery, Lund University and Skåne University Hospital, Lund, Sweden
| | - Stig Steen
- Department of Cardiothoracic Surgery, Lund University and Skåne University Hospital, Lund, Sweden
| | - James E Fildes
- The Ex-Vivo Lab, Division of Cell Matrix and Regenerative Medicine, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, School of Biological Sciences, The University of Manchester, Manchester, United Kingdom.,The Transplant Centre, Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom
| |
Collapse
|
24
|
Moui A, Klein M, Hassoun D, Dijoux E, Cheminant MA, Magnan A, Bouchaud G. The IL-15 / sIL-15Rα complex modulates immunity without effect on asthma features in mouse. Respir Res 2020; 21:33. [PMID: 31996218 PMCID: PMC6988344 DOI: 10.1186/s12931-020-1301-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 01/20/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Interleukin 15 (IL-15) is a growth and modulating factor for B, T lymphocytes and natural killer cells (NK). Its action on innate and adaptive immunity is modulated by its alpha chain receptor (IL-15Rα). The IL-15/sIL-15Rα complex (IL-15Cx) increases the bioavailability and activity of the cytokine in vivo. IL-15Cx has been used in diseases to dampen IL-15 inflammation by the use of soluble IL-15Ralpha specificity. Here, we aim to evaluate the interest of IL-15Cx in a mouse model of asthma. METHODS Using a mouse model of asthma consisting in percutaneous sensitization and intranasal challenge with total house dust mite extract, we evaluated the effect of IL-15Cx injected intraperitoneally four times after a first nasal challenge. Respiratory function was assessed by the technique of forced oscillations (Flexivent®). The effect on bronchial remodeling was evaluated by lung histology. The inflammatory status was analyzed by flow cytometry. RESULTS We observed that the IL-15Cx modulates lung and systemic inflammation by increasing NK cells, CD8+ memory T cells and regulatory cells. However, IL-15Cx displays no effect on bronchial hyperreactivity, bronchial remodeling nor cellular bronchial infiltrate, but limits the secretion of bronchial mucus and modulates only inflammatory response in a HDM-allergic asthma murine model. CONCLUSIONS IL-15Cx has a limited effect on immune response in asthma and has no effect on lung function in mice. Thus, it limits its therapeutic potential but might suggest a combinatory potential with other therapeutics.
Collapse
Affiliation(s)
- Antoine Moui
- L'institut du thorax, Inserm, CNRS, Université́ de Nantes, Nantes, France.,L'institut du thorax, CHU de Nantes, service de pneumologie, Nantes, France
| | - Martin Klein
- L'institut du thorax, Inserm, CNRS, Université́ de Nantes, Nantes, France
| | - Dorian Hassoun
- L'institut du thorax, Inserm, CNRS, Université́ de Nantes, Nantes, France.,L'institut du thorax, CHU de Nantes, service de pneumologie, Nantes, France
| | - Eléonore Dijoux
- L'institut du thorax, Inserm, CNRS, Université́ de Nantes, Nantes, France
| | | | - Antoine Magnan
- L'institut du thorax, Inserm, CNRS, Université́ de Nantes, Nantes, France.,L'institut du thorax, CHU de Nantes, service de pneumologie, Nantes, France
| | - Grégory Bouchaud
- INRA, UR1268 BIA, rue de la Géraudière, F-44316, Nantes, France.
| |
Collapse
|
25
|
Jang JH, Janker F, De Meester I, Arni S, Borgeaud N, Yamada Y, Gil Bazo I, Weder W, Jungraithmayr W. The CD26/DPP4-inhibitor vildagliptin suppresses lung cancer growth via macrophage-mediated NK cell activity. Carcinogenesis 2019; 40:324-334. [PMID: 30698677 DOI: 10.1093/carcin/bgz009] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 11/16/2018] [Accepted: 01/22/2019] [Indexed: 12/20/2022] Open
Abstract
CD26/dipeptidyl peptidase 4 (DPP4) is a transmembrane protein which is expressed by various malignant cells. We found that the expression of CD26/DPP4 was significantly higher in lung adenocarcinoma samples in our own patient cohort compared to normal lung tissue. We therefore hypothesize that the inhibition of CD26/DPP4 can potentially suppress lung cancer growth. The CD26/DPP4 inhibitor vildagliptin was employed on Lewis Lung Carcinoma (LLC) cell line and a human lung adenocarcinoma (H460) cell line. Two weeks after subcutaneous injection of tumor cells into C57BL/6 and CD1/nude mice, the size of LLC and H460 tumors was significantly reduced by vildagliptin. Immunohistochemically, the number of macrophages (F4/80+) and NK cells (NKp46+) was significantly increased in vildagliptin-treated tumor samples. Mechanistically, we found in vitro that lung cancer cell lines expressed increased levels of surfactant protein upon vildagliptin treatment thereby promoting the pro-inflammatory activity of macrophages. By the depletion of macrophages with clodronate and by using NK cell deficient (IL-15-/-) mice, tumors reversed to the size of controls, suggesting that indeed macrophages and NK cells were responsible for the observed tumor-suppressing effect upon vildagliptin treatment. FACS analysis showed tumor-infiltrating NK cells to express tumor necrosis-related apoptosis-inducing ligand (TRAIL) which induced the intra-cellular stress marker γH2AX. Accordingly, we found upregulated γH2AX in vildagliptin-treated tumors and TRAIL-treated cell lines. Moreover, the effect of vildagliptin-mediated enhanced NK cell cytotoxicity could be reversed by antagonizing the TRAIL receptor. Our data provide evidence that the CD26/DPP4-inhibitor vildagliptin reduces lung cancer growth. We could demonstrate that this effect is exerted by surfactant-activated macrophages and NK cells that act against the tumor via TRAIL-mediated cytotoxicity.
Collapse
Affiliation(s)
- Jae-Hwi Jang
- Department of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Florian Janker
- Department of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Ingrid De Meester
- Department of Medical Biochemistry, University of Antwerp, Antwerp, Belgium
| | - Stephan Arni
- Department of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Nathalie Borgeaud
- Department of Visceral Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Yoshito Yamada
- Department of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Ignacio Gil Bazo
- Department of Oncology, University Hospital Navarra, Pamplona, Spain
| | - Walter Weder
- Department of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Wolfgang Jungraithmayr
- Department of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland.,Department of Thoracic Surgery, University Hospital Rostock, Rostock, Germany
| |
Collapse
|
26
|
Calabrese DR, Lanier LL, Greenland JR. Natural killer cells in lung transplantation. Thorax 2018; 74:397-404. [PMID: 30381399 DOI: 10.1136/thoraxjnl-2018-212345] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 09/06/2018] [Accepted: 10/01/2018] [Indexed: 12/16/2022]
Abstract
Natural killer (NK) cells are innate lymphoid cells that have been increasingly recognised as important in lung allograft tolerance and immune defence. These cells evolved to recognise alterations in self through a diverse set of germline-encoded activating and inhibitory receptors and display a broad range of effector functions that play important roles in responding to infections, malignancies and allogeneic tissue. Here, we review NK cells, their diverse receptors and the mechanisms through which NK cells are postulated to mediate important lung transplant clinical outcomes. NK cells can promote tolerance, such as through the depletion of donor antigen-presenting cells. Alternatively, these cells can drive rejection through cytotoxic effects on allograft tissue recognised as 'non-self' or 'stressed', via killer cell immunoglobulin-like receptor (KIR) or NKG2D receptor ligation, respectively. NK cells likely mediate complement-independent antibody-mediated rejection of allografts though CD16A Fc receptor-dependent activation induced by graft-specific antibodies. Finally, NK cells play an important role in response to infections, particularly by mediating cytomegalovirus infection through the CD94/NKG2C receptor. Despite these sometimes-conflicting effects on allograft function, enumeration of NK cells may have an important role in diagnosing allograft dysfunction. While the effects of immunosuppression agents on NK cells may currently be largely unintentional, further understanding of NK cell biology in lung allograft recipients may allow these cells to serve as biomarkers of graft injury and as therapeutic targets.
Collapse
Affiliation(s)
- Daniel R Calabrese
- Department of Medicine, University of California, San Francisco, California, USA
| | - Lewis L Lanier
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, California, USA.,The Parker Institute for Cancer Immunotherapy, University of California San Francisco, San Francisco, California, USA
| | - John R Greenland
- Department of Medicine, University of California, San Francisco, California, USA.,Medical Service, Veterans Affairs Health Care System, San Francisco, California, USA
| |
Collapse
|
27
|
Chronic Airway Fibrosis in Orthotopic Mouse Lung Transplantation Models—An Experimental Reappraisal. Transplantation 2018; 102:e49-e58. [DOI: 10.1097/tp.0000000000001917] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
28
|
Kawakami T, Ito K, Matsuda Y, Noda M, Sakurada A, Hoshikawa Y, Okada Y, Ogasawara K. Cytotoxicity of Natural Killer Cells Activated Through NKG2D Contributes to the Development of Bronchiolitis Obliterans in a Murine Heterotopic Tracheal Transplant Model. Am J Transplant 2017; 17:2338-2349. [PMID: 28251796 DOI: 10.1111/ajt.14257] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 01/26/2017] [Accepted: 02/17/2017] [Indexed: 01/25/2023]
Abstract
Bronchiolitis obliterans after lung transplantation is a major cause of postoperative mortality in which T cell-mediated immunity is known to play an important role. However, the exact contribution of natural killer (NK) cells, which have functions similar to CD8+ T cells, has not been defined. Here, we assessed the role of NK cells in murine bronchiolitis obliterans through heterotopic tracheal transplantations and found a greater percentage of NK cells in allografts than in isografts. Depletion of NK cells using an anti-NK1.1 antibody attenuated bronchiolitis obliterans in transplant recipients compared with controls. In terms of NK cell effector functions, an improvement in bronchiolitis obliterans was observed in perforin-KO recipient mice compared to wild type (WT). Furthermore, we found upregulation of NKG2D-ligand in allografts and demonstrated the significance of this using grafts expressing Rae-1, a murine NKG2D-ligand, which induced severe bronchiolitis obliterans in WT and Rag-1 KO recipients. This effect was ameliorated by injection of anti-NKG2D blocking antibody. Together, these results suggest that cytotoxicity resulting from activation of NK cells through NKG2D leads to the development of murine bronchiolitis obliterans.
Collapse
Affiliation(s)
- T Kawakami
- Department of Immunobiology, Institute of Development, Aging and Cancer, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan.,Department of Thoracic Surgery, Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - K Ito
- Department of Immunobiology, Institute of Development, Aging and Cancer, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Y Matsuda
- Department of Thoracic Surgery, Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - M Noda
- Department of Thoracic Surgery, Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - A Sakurada
- Department of Thoracic Surgery, Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Y Hoshikawa
- Department of Thoracic Surgery, Graduate School of Medicine, Fujita Health University, Toyoake, Aichi, Japan
| | - Y Okada
- Department of Thoracic Surgery, Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - K Ogasawara
- Department of Immunobiology, Institute of Development, Aging and Cancer, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| |
Collapse
|
29
|
Greenland JR, Sun H, Calabrese D, Chong T, Singer JP, Kukreja J, Hays SR, Golden JA, Caughey GH, Venstrom JM, Rajalinginam R. HLA Mismatching Favoring Host-Versus-Graft NK Cell Activity Via KIR3DL1 Is Associated With Improved Outcomes Following Lung Transplantation. Am J Transplant 2017; 17:2192-2199. [PMID: 28375571 PMCID: PMC5519429 DOI: 10.1111/ajt.14295] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 03/07/2017] [Accepted: 03/24/2017] [Indexed: 01/25/2023]
Abstract
Chronic lung allograft dysfunction (CLAD) is linked to rejection and limits survival following lung transplantation. HLA-Bw4 recipients of HLA-Bw6 grafts have enhanced host-versus-graft (HVG) natural killer (NK) cell activity mediated by killer cell immunoglobulin-like receptor (KIR)3DL1 ligand. Because NK cells may promote tolerance by depleting antigen-presenting cells, we hypothesized improved outcomes for HLA-Bw4 recipients of HLA-Bw6 grafts. We evaluated differences in acute cellular rejection and CLAD-free survival across 252 KIR3DL1+ recipients from University of California, San Francisco (UCSF). For validation, we assessed survival and freedom from bronchiolitis obliterans syndrome (BOS), retransplantation, or death in 12 845 non-KIR typed recipients from the United Network for Organ Sharing (UNOS) registry. Cox proportional hazards models were adjusted for age, gender, ethnicity, transplant type, and HLA mismatching. HVG-capable subjects in the UCSF cohort had a decreased risk of CLAD or death (hazard ratio [HR] 0.57, 95% confidence interval [CI] 0.36-0.88) and decreased early lymphocytic bronchitis. The HVG effect was not significant in subjects with genotypes predicting low KIR3DL1 expression. In the UNOS cohort, HVG-capable subjects had a decreased risk of BOS, retransplant, or death (HR 0.95, 95% CI 0.91-0.99). Survival improved with the higher-affinity Bw4-80I ligand and in Bw4 homozygotes. Improved outcomes in HVG-capable recipients are consistent with a protective NK cell role. Augmentation of NK activity could supplement current immunosuppression techniques.
Collapse
Affiliation(s)
- John R. Greenland
- Medical Service, Veterans Affairs Medical Center, San Francisco CA, 94121,Department of Medicine, University of California, San Francisco CA, 94143,Corresponding author:
| | - Haibo Sun
- Immunogenetics and Transplantation Laboratory, Department of Surgery, University of California, San Francisco CA, 94143
| | - Daniel Calabrese
- Department of Medicine, University of California, San Francisco CA, 94143
| | - Tiffany Chong
- Department of Medicine, University of California, San Francisco CA, 94143
| | - Jonathan P. Singer
- Department of Medicine, University of California, San Francisco CA, 94143
| | - Jasleen Kukreja
- Department of Surgery, University of California, San Francisco CA, 94143
| | - Steven R. Hays
- Department of Medicine, University of California, San Francisco CA, 94143
| | - Jeffrey A. Golden
- Department of Medicine, University of California, San Francisco CA, 94143,Department of Surgery, University of California, San Francisco CA, 94143
| | - George H. Caughey
- Medical Service, Veterans Affairs Medical Center, San Francisco CA, 94121,Department of Medicine, University of California, San Francisco CA, 94143,Cardiovascular Research Institute, University of California, San Francisco CA, 94143
| | | | - Raja Rajalinginam
- Immunogenetics and Transplantation Laboratory, Department of Surgery, University of California, San Francisco CA, 94143
| |
Collapse
|
30
|
Jang JH, Yamada Y, Janker F, De Meester I, Baerts L, Vliegen G, Inci I, Chatterjee S, Weder W, Jungraithmayr W. Anti-inflammatory effects on ischemia/reperfusion-injured lung transplants by the cluster of differentiation 26/dipeptidylpeptidase 4 (CD26/DPP4) inhibitor vildagliptin. J Thorac Cardiovasc Surg 2017; 153:713-724.e4. [DOI: 10.1016/j.jtcvs.2016.10.080] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 10/25/2016] [Accepted: 10/27/2016] [Indexed: 12/24/2022]
|
31
|
Hsiao HM, Scozzi D, Gauthier JM, Kreisel D. Mechanisms of graft rejection after lung transplantation. Curr Opin Organ Transplant 2017; 22:29-35. [PMID: 27861263 PMCID: PMC5443682 DOI: 10.1097/mot.0000000000000371] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
PURPOSE OF REVIEW To date, outcomes after lung transplantation are far worse than after transplantation of other solid organs. New insights into mechanisms that contribute to graft rejection and tolerance after lung transplantation remain of great interest. This review examines the recent literature on the role of innate and adaptive immunity in shaping the fate of lung grafts. RECENT FINDINGS Innate and adaptive immune cells orchestrate allograft rejection after transplantation. Innate immune cells such as neutrophils are recruited to the lung graft early after reperfusion and subsequently promote allograft rejection. Although it is widely recognized that CD4 T lymphocytes in concert with CD8 T cells promote graft rejection, regulatory Foxp3 CD4 T, central memory CD8 T cells, and natural killer cells can facilitate tolerance. SUMMARY This review highlights interactions between innate and adaptive immune pathways and how they contribute to lung allograft rejection. These findings lay a foundation for the design of new therapeutic strategies that target both innate and adaptive immune responses.
Collapse
Affiliation(s)
- Hsi-Min Hsiao
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO
| | - Davide Scozzi
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Jason M. Gauthier
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO
| | - Daniel Kreisel
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO
- Department of Pathology & Immunology, Washington University School of Medicine, Saint Louis, MO
| |
Collapse
|
32
|
|
33
|
Diagnostic value of plasma and bronchoalveolar lavage samples in acute lung allograft rejection: differential cytology. Respir Res 2016; 17:74. [PMID: 27323950 PMCID: PMC4915079 DOI: 10.1186/s12931-016-0391-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 06/13/2016] [Indexed: 12/21/2022] Open
Abstract
Diagnosis of acute lung allograft rejection is currently based on transbronchial lung biopsies. Additional methods to detect acute allograft dysfunction derived from plasma and bronchoalveolar lavage samples might facilitate diagnosis and ultimately improve allograft survival. This review article gives an overview of the cell profiles of bronchoalveolar lavage and plasma samples during acute lung allograft rejection. The value of these cells and changes within the pattern of differential cytology to support the diagnosis of acute lung allograft rejection is discussed. Current findings on the topic are highlighted and trends for future research are identified.
Collapse
|
34
|
Stone JP, Critchley WR, Major T, Rajan G, Risnes I, Scott H, Liao Q, Wohlfart B, Sjöberg T, Yonan N, Steen S, Fildes JE. Altered Immunogenicity of Donor Lungs via Removal of Passenger Leukocytes Using Ex Vivo Lung Perfusion. Am J Transplant 2016; 16:33-43. [PMID: 26366523 DOI: 10.1111/ajt.13446] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 06/23/2015] [Accepted: 07/08/2015] [Indexed: 01/25/2023]
Abstract
Passenger leukocyte transfer from the donor lung to the recipient is intrinsically involved in acute rejection. Direct presentation of alloantigen expressed on donor leukocytes is recognized by recipient T cells, promoting acute cellular rejection. We utilized ex vivo lung perfusion (EVLP) to study passenger leukocyte migration from donor lungs into the recipient and to evaluate the effects of donor leukocyte depletion prior to transplantation. For this purpose, female pigs received male left lungs either following 3 h of EVLP or retrieved using standard protocols. Recipients were monitored for 24 h and sequential samples were collected. EVLP-reduced donor leukocyte transfer into the recipient and migration to recipient lymph nodes was markedly reduced. Recipient T cell infiltration of the donor lung was significantly diminished via EVLP. Donor leukocyte removal during EVLP reduces direct allorecognition and T cell priming, diminishing recipient T cell infiltration, the hallmark of acute rejection.
Collapse
Affiliation(s)
- J P Stone
- The Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, UK.,The Transplant Centre, University Hospital of South Manchester, Manchester, UK
| | - W R Critchley
- The Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, UK.,The Transplant Centre, University Hospital of South Manchester, Manchester, UK
| | - T Major
- The Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, UK.,The Transplant Centre, University Hospital of South Manchester, Manchester, UK
| | - G Rajan
- The Transplant Centre, University Hospital of South Manchester, Manchester, UK
| | - I Risnes
- Department of Pathology, Institute of Clinical Medicine, University of Oslo, Rikshospitalet, Oslo, Norway
| | - H Scott
- Department of Pathology, Institute of Clinical Medicine, University of Oslo, Rikshospitalet, Oslo, Norway
| | - Q Liao
- Department of Cardiothoracic Surgery, Lund University and Skåne University Hospital, Lund, Sweden
| | - B Wohlfart
- Department of Cardiothoracic Surgery, Lund University and Skåne University Hospital, Lund, Sweden
| | - T Sjöberg
- Department of Cardiothoracic Surgery, Lund University and Skåne University Hospital, Lund, Sweden
| | - N Yonan
- The Transplant Centre, University Hospital of South Manchester, Manchester, UK
| | - S Steen
- Department of Cardiothoracic Surgery, Lund University and Skåne University Hospital, Lund, Sweden
| | - J E Fildes
- The Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, UK.,The Transplant Centre, University Hospital of South Manchester, Manchester, UK
| |
Collapse
|
35
|
Yamada Y, Jang JH, De Meester I, Baerts L, Vliegen G, Inci I, Yoshino I, Weder W, Jungraithmayr W. CD26 costimulatory blockade improves lung allograft rejection and is associated with enhanced interleukin-10 expression. J Heart Lung Transplant 2015; 35:508-17. [PMID: 26755203 DOI: 10.1016/j.healun.2015.11.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Revised: 10/15/2015] [Accepted: 11/19/2015] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND The ectoenzyme CD26/dipeptidyl peptidase 4 (DPP4) has costimulatory activity that contributes to T cell activation and proliferation. Here, we aimed to target this costimulatory activity for the attenuation of the alloreactive Th17-cell response during acute rejection after mouse lung transplantation. METHODS To test the CD26-costimulatory blockade in vitro, mixed lymphocyte reaction was performed between major histocompatibility complex class I and II fully mismatched cells (CD4(+) splenocytes, C57BL/6, responders, and antigen-presenting cells, BALB/c, stimulators) by adding the CD26 inhibitor vildagliptin (0-15 μg). Lung transplantation between BALB/c (donor) and C57BL/6 (recipient) mice was performed, including controls, CD26-inhibited (CD26-I, daily administration of vildagliptin [GLSynthesis, Worcester, MA], 10 mg/kg subcutaneous), and CD26 knockout (CD26KO) mice was performed. Analysis on Day 1 and 5 after transplant included immunohistochemistry, fluorescence-activated cell sorting, and enzyme-linked immunosorbent assay (ELISA) for immune cell detection and their key cytokines. RESULTS In vitro, there was a significant reduction of the Th17 cytokines interleukin (IL)-17 and IL-21. In vivo, CD26-I-treated and CD26KO mice showed significantly preserved macroscopic and histologic characteristics on Day 5 (p < 0.01), a higher partial pressure of arterial oxygen/fraction of inspired oxygen ratio (p ≤ 0.05), fewer infiltrating CD3(+) T cells (p < 0.01), but more interstitial macrophages on Day 1 (p < 0.01) compared with control. Fewer IL-17(+) cells were found in CD26-I allografts on Day 1 (p = 0.05). Higher levels of IL-10 in CD26-I and CD26KO allografts on day 5 were seen (p < 0.05). IL-10/CD206 double-staining (alternative macrophages) revealed more positive cells in CD26-I and CD26KO on Day 1 and 5 (p < 0.01). CONCLUSIONS CD26 costimulatory blockade promotes lung allograft acceptance via reduced T cell infiltration, less expression of IL-17, and increased expression of IL-10, likely to be derived from alternatively activated macrophages.
Collapse
Affiliation(s)
- Yoshito Yamada
- Division of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland; Department of General Thoracic Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Jae-Hwi Jang
- Division of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Ingrid De Meester
- Department of Medical Biochemistry, University of Antwerp, Antwerp, Belgium
| | - Lesley Baerts
- Department of Medical Biochemistry, University of Antwerp, Antwerp, Belgium
| | - Gwendolyn Vliegen
- Department of Medical Biochemistry, University of Antwerp, Antwerp, Belgium
| | - Ilhan Inci
- Division of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Ichiro Yoshino
- Department of General Thoracic Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Walter Weder
- Division of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | | |
Collapse
|
36
|
Abstract
Research in pulmonary transplantation is actively evolving in quality and scope to meet the challenges of a growing population of lung allograft recipients. In 2013, research groups leveraged large publicly available datasets in addition to multicenter research networks and single-center studies to make significant contributions to our knowledge and clinical care in the areas of donor use, clinical transplant outcomes, mechanisms of rejection, infectious complications, and chronic allograft dysfunction.
Collapse
Affiliation(s)
- Jamie L Todd
- 1 Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | | | | |
Collapse
|
37
|
Greenland JR, Jewell NP, Gottschall M, Trivedi NN, Kukreja J, Hays SR, Singer JP, Golden JA, Caughey GH. Bronchoalveolar lavage cell immunophenotyping facilitates diagnosis of lung allograft rejection. Am J Transplant 2014; 14:831-40. [PMID: 24512389 PMCID: PMC4300200 DOI: 10.1111/ajt.12630] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Revised: 10/21/2013] [Accepted: 10/25/2013] [Indexed: 01/25/2023]
Abstract
Supplementary methods to identify acute rejection and to distinguish rejection from infection may improve clinical outcomes for lung allograft recipients. We hypothesized that distinct bronchoalveolar lavage (BAL) cell profiles are associated with rejection and infection. We retrospectively compared 2939 BAL cell counts and immunophenotypes against concomitantly obtained transbronchial biopsies and microbiologic studies. We randomly assigned 317 subjects to a derivation or validation cohort. BAL samples were classified into four groups: infection, rejection grade ≥A1, both or neither. We employed generalized estimating equation and survival modeling to identify clinical predictors of rejection and infection. We found that CD25(+) and natural killer cell percentages identified a twofold increased odds of rejection compared to either the infection or the neither infection nor rejection groups. Also, monocytes, lymphocytes and eosinophil percentages were independently associated with rejection. A four-predictor scoring system had high negative predictive value (96-98%) for grade ≥A2 rejection, predicted future rejection in the validation cohort and predicted increased risk of bronchiolitis obliterans syndrome in otherwise benign samples. In conclusion, BAL cell immunophenotyping discriminates between infection and acute rejection and predicts future outcomes in lung transplant recipients. Although it cannot replace histopathology, immunophenotyping may be a clinically useful adjunct.
Collapse
Affiliation(s)
- JR Greenland
- Department of Medicine, University of California at San Francisco, CA 94143,Medical Service, Veterans Affairs Medical Center, San Francisco, CA 94121
| | - NP Jewell
- Division of Biostatistics, University of California, Berkeley, CA, 94720
| | - M Gottschall
- Department of Pathology, University of California at San Francisco, CA 94143
| | - NN Trivedi
- Department of Medicine, University of California at San Francisco, CA 94143,Medical Service, Veterans Affairs Medical Center, San Francisco, CA 94121
| | - J Kukreja
- Department of Surgery, University of California at San Francisco, CA 94143
| | - SR Hays
- Department of Medicine, University of California at San Francisco, CA 94143
| | - JP Singer
- Department of Medicine, University of California at San Francisco, CA 94143
| | - JA Golden
- Department of Medicine, University of California at San Francisco, CA 94143
| | - GH Caughey
- Department of Medicine, University of California at San Francisco, CA 94143,Cardiovascular Research Institute, University of California at San Francisco, CA 94143,Medical Service, Veterans Affairs Medical Center, San Francisco, CA 94121
| |
Collapse
|
38
|
Abstract
PURPOSE OF REVIEW To examine the recent literature on the role of innate cells in immunity to transplanted tissue. It specifically addresses the impact of monocytes/macrophages, neutrophils, natural killer cells, and platelets. RECENT FINDINGS Current research indicates that innate immunity plays a dual role in response to transplanted tissue with the ability to either facilitate rejection or promote tolerance. Intriguingly, some of these cells are even capable of reacting to allogeneic cells, a feature usually only attributed to cells of the adaptive immune system. SUMMARY This review highlights the new therapeutic targets in the innate immune system that may be useful in the treatment of transplant recipients. It also emphasizes the need to use caution in exploring these new therapeutics.
Collapse
|
39
|
|
40
|
Maguire O, Tario JD, Shanahan TC, Wallace PK, Minderman H. Flow cytometry and solid organ transplantation: a perfect match. Immunol Invest 2014; 43:756-74. [PMID: 25296232 PMCID: PMC4357273 DOI: 10.3109/08820139.2014.910022] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In the field of transplantation, flow cytometry serves a well-established role in pre-transplant crossmatching and monitoring immune reconstitution following hematopoietic stem cell transplantation. The capabilities of flow cytometers have continuously expanded and this combined with more detailed knowledge of the constituents of the immune system, their function and interaction and newly developed reagents to study these parameters have led to additional utility of flow cytometry-based analyses, particularly in the post-transplant setting. This review discusses the impact of flow cytometry on managing alloantigen reactions, monitoring opportunistic infections and graft rejection and gauging immunosuppression in the context of solid organ transplantation.
Collapse
Affiliation(s)
- Orla Maguire
- Laboratory of Flow and Image Cytometry, Department of Pathology and Laboratory Medicine, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Joseph D. Tario
- Laboratory of Flow and Image Cytometry, Department of Pathology and Laboratory Medicine, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Thomas C. Shanahan
- Department of Microbiology and Immunology, State University of New York at Buffalo, Buffalo, New York, USA
| | - Paul K. Wallace
- Laboratory of Flow and Image Cytometry, Department of Pathology and Laboratory Medicine, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Hans Minderman
- Laboratory of Flow and Image Cytometry, Department of Pathology and Laboratory Medicine, Roswell Park Cancer Institute, Buffalo, New York, USA
| |
Collapse
|
41
|
Chijioke O, Münz C. Dendritic cell derived cytokines in human natural killer cell differentiation and activation. Front Immunol 2013; 4:365. [PMID: 24273539 PMCID: PMC3822368 DOI: 10.3389/fimmu.2013.00365] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Accepted: 10/27/2013] [Indexed: 12/16/2022] Open
Abstract
Dendritic cells (DCs) and natural killer (NK) cells shape each other’s functions early during immune responses. DCs activate NK cells and NK cells can mature or kill DCs. In this review we will discuss which DC and NK cell subsets are mainly affected by this interaction, where these encounters might take place and which signals are exchanged. Finally, we will point out what the clinical benefit of understanding this interaction might be and how it changed our view on NK cells as innate lymphocytes.
Collapse
Affiliation(s)
- Obinna Chijioke
- Viral Immunobiology, Institute of Experimental Immunology, University of Zurich , Zurich , Switzerland
| | | |
Collapse
|
42
|
Shilling RA. Harnessing natural killer cells to protect lung transplants from acute rejection. Am J Respir Crit Care Med 2013; 187:1284-6. [PMID: 23767901 DOI: 10.1164/rccm.201304-0634ed] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|